1
|
Askari K, Johnson JL, Shukla A, Meneses-Salas E, Kiosses WB, Yu J, Catz SD. Nexinhib20 inhibits JFC1-mediated mobilization of a subset of CD11b/CD18+ vesicles decreasing integrin avidity, but does not inhibit Rac1. J Leukoc Biol 2025; 117:qiaf012. [PMID: 39883854 PMCID: PMC12022635 DOI: 10.1093/jleuko/qiaf012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 01/02/2025] [Accepted: 01/28/2025] [Indexed: 02/01/2025] Open
Abstract
Regulated sequential exocytosis of neutrophil granules is essential for orchestrating the innate immune response, while uncontrolled secretion causes inflammation. We developed and characterized Nexinhib20, a small-molecule inhibitor that targets azurophilic granule exocytosis in neutrophils by blocking the interaction between the small GTPase Rab27a and its effector JFC1. Its therapeutic potential has been demonstrated in several preclinical models of inflammatory disease. Here, using neutrophils from Jfc1-KO mice, we show that JFC1 regulates the mobilization of a small subpopulation of CD11b+ granules. Nexinhib20 inhibits the mobilization of β2-integrins from a subset of CD11b+ granules to the plasma membrane in human and mouse neutrophils. The putative impact of Nexinhib20 on integrin activation is caused by decreased avidity, secondary to its effect on β2-integrin mobilization. CD11b mobilization and integrin activation were unaffected by pharmacological inhibition or activation of Rac1. Using quantitative 3D enhanced resolution microscopy, we show that neutrophil activation induces the recruitment of JFC1 to CD11b+ granules. Nexinhib20 decreased the localization of JFC1 at CD11b+ granules without affecting the association of Rac1 with CD11b. Nexinhib20 inhibits JFC1 recruitment but not endogenous Rac1 activation in living cells. Using orthogonal analyses of Rac1 activity consisting of a sensitive, time-resolved fluorescence energy transfer, Rac1-PAK1-binding assay, and endogenous Rac1-GTP examination, we show that Nexinhib20 does not interfere with Rac1 activation. Instead, we confirm its molecular mode of action as the inhibition of the Rab27a-JFC1 binding. Thus, Nexinhib20 limits β2-integrin mobilization to the cell surface, decreasing avidity and affecting active integrin availability in a JFC1-dependent but Rac1-independent manner.
Collapse
Affiliation(s)
- Kasra Askari
- Department of Molecular and Cellular Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, United States
| | - Jennifer L Johnson
- Department of Molecular and Cellular Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, United States
| | - Aparna Shukla
- Department of Molecular and Cellular Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, United States
| | - Elsa Meneses-Salas
- Department of Molecular and Cellular Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, United States
| | - William B Kiosses
- Division of Inflammation Biology, La Jolla Institute for Immunology, 9420 Athena Cir, La Jolla, CA 92037, United States
| | - Juan Yu
- Department of Molecular and Cellular Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, United States
| | - Sergio D Catz
- Department of Molecular and Cellular Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, United States
| |
Collapse
|
2
|
Zhong W, Lu Y, Han X, Yang J, Qin Z, Zhang W, Yu Z, Wu B, Liu S, Xu W, Zheng C, Schuchter LM, Karakousis GC, Mitchell TC, Amaravadi R, Flowers AJ, Gimotty PA, Xiao M, Mills G, Herlyn M, Dong H, Mitchell MJ, Kim J, Xu X, Guo W. Upregulation of exosome secretion from tumor-associated macrophages plays a key role in the suppression of anti-tumor immunity. Cell Rep 2023; 42:113224. [PMID: 37805922 PMCID: PMC10697782 DOI: 10.1016/j.celrep.2023.113224] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 06/15/2023] [Accepted: 09/20/2023] [Indexed: 10/10/2023] Open
Abstract
Macrophages play a pivotal role in tumor immunity. We report that reprogramming of macrophages to tumor-associated macrophages (TAMs) promotes the secretion of exosomes. Mechanistically, increased exosome secretion is driven by MADD, which is phosphorylated by Akt upon TAM induction and activates Rab27a. TAM exosomes carry high levels of programmed death-ligand 1 (PD-L1) and potently suppress the proliferation and function of CD8+ T cells. Analysis of patient melanoma tissues indicates that TAM exosomes contribute significantly to CD8+ T cell suppression. Single-cell RNA sequencing analysis showed that exosome-related genes are highly expressed in macrophages in melanoma; TAM-specific RAB27A expression inversely correlates with CD8+ T cell infiltration. In a murine melanoma model, lipid nanoparticle delivery of small interfering RNAs (siRNAs) targeting macrophage RAB27A led to better T cell activation and sensitized tumors to anti-programmed cell death protein 1 (PD-1) treatment. Our study demonstrates tumors use TAM exosomes to combat CD8 T cells and suggests targeting TAM exosomes as a potential strategy to improve immunotherapies.
Collapse
Affiliation(s)
- Wenqun Zhong
- Department of Biology, School of Arts & Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Youtao Lu
- Department of Biology, School of Arts & Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Xuexiang Han
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jingbo Yang
- Department of Biology, School of Arts & Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Zhiyuan Qin
- Department of Biology, School of Arts & Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Wei Zhang
- Department of Biology, School of Arts & Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ziyan Yu
- Department of Biology, School of Arts & Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Bin Wu
- Department of Biology, School of Arts & Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Shujing Liu
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Wei Xu
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Cathy Zheng
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Lynn M Schuchter
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Giorgos C Karakousis
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Tara C Mitchell
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ravi Amaravadi
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ahron J Flowers
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Phyllis A Gimotty
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Min Xiao
- Molecular and Cellular Oncogenesis Program and Melanoma Research Center, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Gordon Mills
- Division of Oncological Science, School of Medicine, and Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97201, USA
| | - Meenhard Herlyn
- Molecular and Cellular Oncogenesis Program and Melanoma Research Center, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Haidong Dong
- Departments of Urology and Immunology, Mayo College of Medicine and Science, Rochester, MN 55905, USA
| | - Michael J Mitchell
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Junhyong Kim
- Department of Biology, School of Arts & Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Xiaowei Xu
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Wei Guo
- Department of Biology, School of Arts & Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
3
|
Meijuan C, Meng X, Fang L, Qian W. Synaptotagmin-like protein 1 is a potential diagnostic and prognostic biomarker in endometrial cancer based on bioinformatics and experiments. J Ovarian Res 2023; 16:16. [PMID: 36653850 PMCID: PMC9850549 DOI: 10.1186/s13048-023-01097-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 01/11/2023] [Indexed: 01/20/2023] Open
Abstract
Endometrial cancer (EC) is one of the most common gynecologic malignancies. Identification of potential EC biomarkers is essential to improve the prognosis and development of therapies against EC. Synaptotagmin-like protein 1 (SYTL1), as a small GTPase Rab27 effector, mainly plays a role in vesicle trafficking and cytotoxic granule exocytosis in lymphocytes. However the role of SYTL1 in EC remains uncertain. We performed a comprehensive assessment of the relationship between SYTL1 and patient diagnosis and prognosis by analysis of EC patients' data from TCGA. We employed the LinkedOmics and Search Tool for the Retrieval of Interacting Genes/Proteins (STRING) database to analyze the biological function of SYTL1 in EC. In addition, the correlation between SYTL1 expression and its DNA methylation was performed by using cBioportal, UALCAN, TCGA Wanderer and MethSurv databases. We further assessed the link between SYTL1 and tumor-infiltrating immune cells by using gene set variation analysis (GSVA).Results We found that SYTL1 was highly expressed in EC patients and cell lines. And increased expression of SYTL1 was associated with age, clinical stage, histological type, histological grade and good overall survival (OS).SYTL1 DNA methylation is negatively associated with SYTL1 expression and UCEC patients' OS. SYTL1 expression is closely correlated with immune infiltration. Furthermore, we carried out in vitro experiments to verify the results of bioinformatic analysis.Conclusion Our results demonstrated that the elevation of SYTL1 expression is associated with good OS and SYTL1 might be a potential diagnostic and prognostic marker in EC.
Collapse
Affiliation(s)
- Cai Meijuan
- grid.452402.50000 0004 1808 3430Department of Clinical Laboratory, Qilu Hospital of Shandong University, No.107 Wenhua West Road, 250013 Jinan, Shandong China ,grid.452402.50000 0004 1808 3430Department of Clinical Laboratory, Qilu Hospital of Shandong University (Qingdao), Qingdao, Shandong China
| | - Xu Meng
- grid.452402.50000 0004 1808 3430Department of Clinical Laboratory, Qilu Hospital of Shandong University (Qingdao), Qingdao, Shandong China
| | - Liu Fang
- Department of Pathology, Qingdao Chengyang People’s Hospital, No.758 Hefei Road, Shandong 266035 Qingdao, China
| | - Wang Qian
- grid.452402.50000 0004 1808 3430Department of Clinical Laboratory, Qilu Hospital of Shandong University, No.107 Wenhua West Road, 250013 Jinan, Shandong China ,grid.452402.50000 0004 1808 3430Department of Clinical Laboratory, Qilu Hospital of Shandong University (Qingdao), Qingdao, Shandong China
| |
Collapse
|
4
|
Liu D, Chen H, Fu Y, Yao Y, He S, Wang Y, Cao Z, Wang X, Yang M, Zhao Q. KCa3.1 Promotes Proinflammatory Exosome Secretion by Activating AKT/Rab27a in Atrial Myocytes during Rapid Pacing. Cardiovasc Ther 2023; 2023:3939360. [PMID: 37035755 PMCID: PMC10079387 DOI: 10.1155/2023/3939360] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 03/04/2023] [Accepted: 03/07/2023] [Indexed: 04/11/2023] Open
Abstract
Purpose The aim of this study was to investigate the role of the medium-conductance calcium-activated potassium channel (KCNN4, KCa3.1) in the secretion of proinflammatory exosomes by atrial myocytes. Methods Eighteen beagles were randomly divided into the sham group (n = 6), pacing group (n = 6), and pacing+TRAM-34 group (n = 6). Electrophysiological data, such as the effective refractory period, atrial fibrillation (AF) induction, and AF duration, were collected by programmed stimulation. Atrial tissues were subjected to hematoxylin and eosin, Masson's trichrome, and immunofluorescence staining. The expression of KCa3.1 and Rab27a was assessed by immunohistochemistry and western blotting. The downstream signaling pathways involved in KCa3.1 were examined by rapid pacing or overexpressing KCNN4 in HL-1 cells. Results Atrial rapid pacing significantly induced electrical remodeling, inflammation, fibrosis, and exosome secretion in the canine atrium, while TRAM-34 (KCa3.1 blocker) inhibited these changes. Compared with those in control HL-1 cells, the levels of exosome markers and inflammatory factors were increased in pacing HL-1 cells. Furthermore, the levels of CD68 and iNOS in macrophages incubated with exosomes derived from HL-1 cells were higher in the pacing-exo group than in the control group. More importantly, KCa3.1 regulated exosome secretion through the AKT/Rab27a signaling pathway. Similarly, inhibiting the downstream signaling pathway of KCa3.1 significantly inhibited exosome secretion. Conclusions KCa3.1 promotes proinflammatory exosome secretion through the AKT/Rab27a signaling pathway. Inhibiting the KCa3.1/AKT/Rab27a signaling pathway reduces myocardial tissue structural remodeling in AF.
Collapse
Affiliation(s)
- Dishiwen Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China
- Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| | - Huiyu Chen
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China
- Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| | - Yuntao Fu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China
- Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| | - Yajun Yao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China
- Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| | - Shanqing He
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China
- Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| | - Youcheng Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China
- Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| | - Zhen Cao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China
- Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| | - Xuewen Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China
- Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| | - Mei Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China
- Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| | - Qingyan Zhao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China
- Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| |
Collapse
|
5
|
Johnson JL, Meneses-Salas E, Ramadass M, Monfregola J, Rahman F, Carvalho Gontijo R, Kiosses WB, Pestonjamasp K, Allen D, Zhang J, Osborne DG, Zhu YP, Wineinger N, Askari K, Chen D, Yu J, Henderson SC, Hedrick CC, Ursini MV, Grinstein S, Billadeau DD, Catz SD. Differential dysregulation of granule subsets in WASH-deficient neutrophil leukocytes resulting in inflammation. Nat Commun 2022; 13:5529. [PMID: 36130971 PMCID: PMC9492659 DOI: 10.1038/s41467-022-33230-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 09/08/2022] [Indexed: 11/09/2022] Open
Abstract
Dysregulated secretion in neutrophil leukocytes associates with human inflammatory disease. The exocytosis response to triggering stimuli is sequential; gelatinase granules modulate the initiation of the innate immune response, followed by the release of pro-inflammatory azurophilic granules, requiring stronger stimulation. Exocytosis requires actin depolymerization which is actively counteracted under non-stimulatory conditions. Here we show that the actin nucleator, WASH, is necessary to maintain azurophilic granules in their refractory state by granule actin entrapment and interference with the Rab27a-JFC1 exocytic machinery. On the contrary, gelatinase granules of WASH-deficient neutrophil leukocytes are characterized by decreased Rac1, shortened granule-associated actin comets and impaired exocytosis. Rac1 activation restores exocytosis of these granules. In vivo, WASH deficiency induces exacerbated azurophilic granule exocytosis, inflammation, and decreased survival. WASH deficiency thus differentially impacts neutrophil granule subtypes, impairing exocytosis of granules that mediate the initiation of the neutrophil innate response while exacerbating pro-inflammatory granule secretion.
Collapse
Affiliation(s)
- Jennifer L Johnson
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Elsa Meneses-Salas
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Mahalakshmi Ramadass
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Jlenia Monfregola
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
- Telethon Institute of Genetics and Medicine (TIGEM), Via Campi Flegrei 34, 80078, Pozzuoli, Naples, Italy
| | - Farhana Rahman
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | | | - William B Kiosses
- Division of Inflammation Biology, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Kersi Pestonjamasp
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Dale Allen
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Jinzhong Zhang
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Douglas G Osborne
- The Division of Oncology Research, Schulze Center for Novel Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Yanfang Peipei Zhu
- Division of Inflammation Biology, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Nathan Wineinger
- Research Translational Institute, Statistics, The Scripps Research Institute, La Jolla, CA, USA
| | - Kasra Askari
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Danni Chen
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Juan Yu
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Scott C Henderson
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Catherine C Hedrick
- Division of Inflammation Biology, La Jolla Institute for Immunology, La Jolla, CA, USA
| | | | - Sergio Grinstein
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| | - Daniel D Billadeau
- The Division of Oncology Research, Schulze Center for Novel Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Sergio D Catz
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA.
| |
Collapse
|
6
|
Chen GH, Yang JG, Xia HF, Zhang LZ, Chen YH, Wang KM, Duan X, Wu LZ, Zhao YF, Chen G. Endothelial cells induce degradation of ECM through enhanced secretion of MMP14 carried on extracellular vesicles in venous malformation. Cell Tissue Res 2022; 389:517-530. [PMID: 35786766 DOI: 10.1007/s00441-022-03657-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Accepted: 06/15/2022] [Indexed: 11/25/2022]
Abstract
Venous malformations (VMs), featuring localized dilated veins, are the most common developmental vascular anomalies. Aberrantly organized perivascular extracellular matrix (ECM) is one of the prominent pathological hallmarks of VMs, accounting for vascular dysfunction. Although previous studies have revealed various proteins involved in ECM remodeling, the detailed pattern and molecular mechanisms underlying the endothelium-ECM interplay have not been fully elucidated. Our previous studies revealed drastically elevated extracellular vesicle (EV) secretion in VM lesions. Here, we identified increased EV-carried MMP14 in lesion fluids of VMs and culture medium of TIE2-L914F mutant endothelial cells (ECs), along with stronger ECM degradation. Knockdown of RAB27A, a required regulator for vesicle docking and fusion, led to decreased secretion of EV-carried MMP14 in vitro. Histochemical analysis further demonstrated a highly positive correlation between RAB27A in the endothelium and MMP14 in the perivascular environment. Therefore, our results proved that RAB27A-regulated secretion of EV-MMP14, as a new pattern of endothelium-ECM interplay, contributed to the development of VMs by promoting ECM degradation.
Collapse
Affiliation(s)
- Gao-Hong Chen
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Jie-Gang Yang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China.,Department of Oral Maxillofacial Head Neck Oncology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Hou-Fu Xia
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China.,Department of Oral Maxillofacial Head Neck Oncology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Lin-Zhou Zhang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Yin-Hsueh Chen
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Kui-Ming Wang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Xu Duan
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Lian-Zhi Wu
- Department of Obstetrics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yi-Fang Zhao
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China.,Department of Oral Maxillofacial Head Neck Oncology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Gang Chen
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China. .,Department of Oral Maxillofacial Head Neck Oncology, School and Hospital of Stomatology, Wuhan University, Wuhan, China. .,Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, China.
| |
Collapse
|
7
|
Ramadass M, Johnson JL, Marki A, Zhang J, Wolf D, Kiosses WB, Pestonjamasp K, Ley K, Catz SD. The trafficking protein JFC1 regulates Rac1-GTP localization at the uropod controlling neutrophil chemotaxis and in vivo migration. J Leukoc Biol 2019; 105:1209-1224. [PMID: 30748033 DOI: 10.1002/jlb.1vma0818-320r] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 01/09/2019] [Accepted: 01/22/2019] [Indexed: 01/01/2023] Open
Abstract
Neutrophil chemotaxis is essential in responses to infection and underlies inflammation. In neutrophils, the small GTPase Rac1 has discrete functions at both the leading edge and in the retraction of the trailing structure at the cell's rear (uropod), but how Rac1 is regulated at the uropod is unknown. Here, we identified a mechanism mediated by the trafficking protein synaptotagmin-like 1 (SYTL1 or JFC1) that controls Rac1-GTP recycling from the uropod and promotes directional migration of neutrophils. JFC1-null neutrophils displayed defective polarization and impaired directional migration to N-formyl-methionine-leucyl-phenylalanine in vitro, but chemoattractant-induced actin remodeling, calcium signaling and Erk activation were normal in these cells. Defective chemotaxis was not explained by impaired azurophilic granule exocytosis associated with JFC1 deficiency. Mechanistically, we show that active Rac1 localizes at dynamic vesicles where endogenous JFC1 colocalizes with Rac1-GTP. Super-resolution microscopy (STORM) analysis shows adjacent distribution of JFC1 and Rac1-GTP, which increases upon activation. JFC1 interacts with Rac1-GTP in a Rab27a-independent manner to regulate Rac1-GTP trafficking. JFC1-null cells exhibited Rac1-GTP accumulation at the uropod and increased tail length, and Rac1-GTP uropod accumulation was recapitulated by inhibition of ROCK or by interference with microtubule remodeling. In vivo, neutrophil dynamic studies in mixed bone marrow chimeric mice show that JFC1-/- neutrophils are unable to move directionally toward the source of the chemoattractant, supporting the notion that JFC1 deficiency results in defective neutrophil migration. Our results suggest that defective Rac1-GTP recycling from the uropod affects directionality and highlight JFC1-mediated Rac1 trafficking as a potential target to regulate chemotaxis in inflammation and immunity.
Collapse
Affiliation(s)
- Mahalakshmi Ramadass
- Department of Molecular Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California, USA
| | - Jennifer L Johnson
- Department of Molecular Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California, USA
| | - Alex Marki
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, California, USA
| | - Jinzhong Zhang
- Department of Molecular Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California, USA
| | - Dennis Wolf
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, California, USA
| | - William B Kiosses
- Department of Molecular Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California, USA
| | - Kersi Pestonjamasp
- Department of Molecular Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California, USA
| | - Klaus Ley
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, California, USA
| | - Sergio D Catz
- Department of Molecular Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California, USA
| |
Collapse
|
8
|
Wahlmüller FC, Sokolikova B, Rieger D, Geiger M. New lipid interaction partners stimulate the inhibition of activated protein C by cell-penetrating protein C inhibitor. Thromb Haemost 2017; 111:41-52. [DOI: 10.1160/th13-06-0478] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Accepted: 08/24/2013] [Indexed: 12/18/2022]
Abstract
SummaryProtein C inhibitor (PCI, SerpinA5) is a heparin-binding serpin which can penetrate through cellular membranes. Selected negatively charged phospholipids like unsaturated phosphatidylserine and oxidised phosphatidylethanolamine bind to PCI and stimulate its inhibitory activity towards different proteases. The interaction of phospholipids with PCI might also alter the lipid distribution pattern of blood cells and influence the remodelling of cellular membranes. Here we showed that PCI is an additional binding partner of phosphatidic acid (PA), cardiolipin (CL), and phosphoinositides (PIPs). Protein lipid overlay assays exhibited a unique binding pattern of PCI towards different lipid species. In addition PA, CL, and unsaturated, monophosphorylated PIPs stimulated the inhibitory property of PCI towards activated protein C in a heparin like manner. As shown for kallistatin (SerpinA4) and vaspin (SerpinA12), the incubation of cells with PCI led to the activation of protein kinase B (AKT), which could be achieved through direct interaction of PCI with PIPs. This model is supported by the fact that PCI stimulated the PIP-dependent 5-phosphatase SHIP2 in vitro, which would result in AKT activation. Hence the interaction of PCI with different lipids might not only stimulate the inhibition of potential target protease by PCI, but could also alter intracellular lipid signalling.
Collapse
|
9
|
McLeish KR, Merchant ML, Creed TM, Tandon S, Barati MT, Uriarte SM, Ward RA. Frontline Science: Tumor necrosis factor-α stimulation and priming of human neutrophil granule exocytosis. J Leukoc Biol 2017; 102:19-29. [PMID: 28096297 DOI: 10.1189/jlb.3hi0716-293rr] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2016] [Revised: 12/19/2016] [Accepted: 12/20/2016] [Indexed: 12/24/2022] Open
Abstract
Neutrophil granule exocytosis plays an important role in innate and adaptive immune responses. The present study examined TNF-α stimulation or priming of exocytosis of the 4 neutrophil granule subsets. TNF-α stimulated exocytosis of secretory vesicles and gelatinase granules and primed specific and azurophilic granule exocytosis to fMLF stimulation. Both stimulation and priming of exocytosis by TNF-α were dependent on p38 MAPK activity. Bioinformatic analysis of 1115 neutrophil proteins identified by mass spectrometry as being phosphorylated by TNF-α exposure found that actin cytoskeleton regulation was a major biologic function. A role for p38 MAPK regulation of the actin cytoskeleton was confirmed experimentally. Thirteen phosphoproteins regulated secretory vesicle quantity, formation, or release, 4 of which-Raf1, myristoylated alanine-rich protein kinase C (PKC) substrate (MARCKS), Abelson murine leukemia interactor 1 (ABI1), and myosin VI-were targets of the p38 MAPK pathway. Pharmacologic inhibition of Raf1 reduced stimulated exocytosis of gelatinase granules and priming of specific granule exocytosis. We conclude that differential regulation of exocytosis by TNF-α involves the actin cytoskeleton and is a necessary component for priming of the 2 major neutrophil antimicrobial defense mechanisms: oxygen radical generation and release of toxic granule contents.
Collapse
Affiliation(s)
- Kenneth R McLeish
- Department of Medicine, University of Louisville School of Medicine, Louisville, Kentucky, USA; and .,Robley Rex Veterans Affairs Medical Center, Louisville, Kentucky, USA
| | - Michael L Merchant
- Department of Medicine, University of Louisville School of Medicine, Louisville, Kentucky, USA; and
| | - T Michael Creed
- Department of Medicine, University of Louisville School of Medicine, Louisville, Kentucky, USA; and
| | - Shweta Tandon
- Department of Medicine, University of Louisville School of Medicine, Louisville, Kentucky, USA; and
| | - Michelle T Barati
- Department of Medicine, University of Louisville School of Medicine, Louisville, Kentucky, USA; and
| | - Silvia M Uriarte
- Department of Medicine, University of Louisville School of Medicine, Louisville, Kentucky, USA; and
| | - Richard A Ward
- Department of Medicine, University of Louisville School of Medicine, Louisville, Kentucky, USA; and
| |
Collapse
|
10
|
Johnson JL, Ramadass M, He J, Brown SJ, Zhang J, Abgaryan L, Biris N, Gavathiotis E, Rosen H, Catz SD. Identification of Neutrophil Exocytosis Inhibitors (Nexinhibs), Small Molecule Inhibitors of Neutrophil Exocytosis and Inflammation: DRUGGABILITY OF THE SMALL GTPase Rab27a. J Biol Chem 2016; 291:25965-25982. [PMID: 27702998 DOI: 10.1074/jbc.m116.741884] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 09/21/2016] [Indexed: 12/22/2022] Open
Abstract
Neutrophils constitute the first line of cellular defense in response to bacterial and fungal infections and rely on granular proteins to kill microorganisms, but uncontrolled secretion of neutrophil cargos is injurious to the host and should be closely regulated. Thus, increased plasma levels of neutrophil secretory proteins, including myeloperoxidase and elastase, are associated with tissue damage and are hallmarks of systemic inflammation. Here, we describe a novel high-throughput screening approach to identify small molecule inhibitors of the interaction between the small GTPase Rab27a and its effector JFC1, two central regulators of neutrophil exocytosis. Using this assay, we have identified small molecule inhibitors of Rab27a-JFC1 binding that were also active in cell-based neutrophil-specific exocytosis assays, demonstrating the druggability of Rab GTPases and their effectors. These compounds, named Nexinhibs (neutrophil exocytosis inhibitors), inhibit exocytosis of azurophilic granules in human neutrophils without affecting other important innate immune responses, including phagocytosis and neutrophil extracellular trap production. Furthermore, the compounds are reversible and potent inhibitors of the extracellular production of superoxide anion by preventing the up-regulation of the granule membrane-associated subunit of the NADPH oxidase at the plasma membrane. Nexinhibs also inhibit the up-regulation of activation signature molecules, including the adhesion molecules CD11b and CD66b. Importantly, by using a mouse model of endotoxin-induced systemic inflammation, we show that these inhibitors have significant activity in vivo manifested by decreased plasma levels of neutrophil secretory proteins and significantly decreased tissue infiltration by inflammatory neutrophils. Altogether, our data present the first neutrophil exocytosis-specific inhibitor with in vivo anti-inflammatory activity, supporting its potential use as an inhibitor of systemic inflammation.
Collapse
Affiliation(s)
| | | | - Jing He
- From the Departments of Molecular and Experimental Medicine and
| | - Steven J Brown
- Chemical Physiology, The Scripps Research Institute, La Jolla, California 92037 and
| | - Jinzhong Zhang
- From the Departments of Molecular and Experimental Medicine and
| | - Lusine Abgaryan
- Chemical Physiology, The Scripps Research Institute, La Jolla, California 92037 and
| | - Nikolaos Biris
- the Departments of Biochemistry and Medicine, Albert Einstein College of Medicine, Bronx, New York 10461
| | - Evripidis Gavathiotis
- the Departments of Biochemistry and Medicine, Albert Einstein College of Medicine, Bronx, New York 10461
| | - Hugh Rosen
- Chemical Physiology, The Scripps Research Institute, La Jolla, California 92037 and
| | - Sergio D Catz
- From the Departments of Molecular and Experimental Medicine and
| |
Collapse
|
11
|
Johnson JL, He J, Ramadass M, Pestonjamasp K, Kiosses WB, Zhang J, Catz SD. Munc13-4 Is a Rab11-binding Protein That Regulates Rab11-positive Vesicle Trafficking and Docking at the Plasma Membrane. J Biol Chem 2015; 291:3423-38. [PMID: 26637356 DOI: 10.1074/jbc.m115.705871] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Indexed: 11/06/2022] Open
Abstract
The small GTPase Rab11 and its effectors control trafficking of recycling endosomes, receptor replenishment and the up-regulation of adhesion and adaptor molecules at the plasma membrane. Despite recent advances in the understanding of Rab11-regulated mechanisms, the final steps mediating docking and fusion of Rab11-positive vesicles at the plasma membrane are not fully understood. Munc13-4 is a docking factor proposed to regulate fusion through interactions with SNAREs. In hematopoietic cells, including neutrophils, Munc13-4 regulates exocytosis in a Rab27a-dependent manner, but its possible regulation of other GTPases has not been explored in detail. Here, we show that Munc13-4 binds to Rab11 and regulates the trafficking of Rab11-containing vesicles. Using a novel Time-resolved Fluorescence Resonance Energy Transfer (TR-FRET) assay, we demonstrate that Munc13-4 binds to Rab11a but not to dominant negative Rab11a. Immunoprecipitation analysis confirmed the specificity of the interaction between Munc13-4 and Rab11, and super-resolution microscopy studies support the interaction of endogenous Munc13-4 with Rab11 at the single molecule level in neutrophils. Vesicular dynamic analysis shows the common spatio-temporal distribution of Munc13-4 and Rab11, while expression of a calcium binding-deficient mutant of Munc13-4 significantly affected Rab11 trafficking. Munc13-4-deficient neutrophils showed normal endocytosis, but the trafficking, up-regulation, and retention of Rab11-positive vesicles at the plasma membrane was significantly impaired. This correlated with deficient NADPH oxidase activation at the plasma membrane in response to Rab11 interference. Our data demonstrate that Munc13-4 is a Rab11-binding partner that regulates the final steps of Rab11-positive vesicle docking at the plasma membrane.
Collapse
Affiliation(s)
| | - Jing He
- From the Department of Molecular and Experimental Medicine and
| | | | - Kersi Pestonjamasp
- Cancer Center Microscopy Shared Resource, University of California San Diego, La Jolla, California 92093
| | - William B Kiosses
- Light Microscopy Core Facility, The Scripps Research Institute, La Jolla, California 92037 and
| | - Jinzhong Zhang
- From the Department of Molecular and Experimental Medicine and
| | - Sergio D Catz
- From the Department of Molecular and Experimental Medicine and
| |
Collapse
|
12
|
Cytokeratin19 induced by HER2/ERK binds and stabilizes HER2 on cell membranes. Cell Death Differ 2014; 22:665-76. [PMID: 25342465 DOI: 10.1038/cdd.2014.155] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2014] [Revised: 08/12/2014] [Accepted: 08/29/2014] [Indexed: 01/22/2023] Open
Abstract
Cytokeratin19 (KRT19) is widely used as a biomarker for the detection of disseminated tumors. Using an LC-MS/MS proteomics approach, we found that KRT19 was upregulated in HER2-overexpressing cells and tissues. KRT19 expression was induced by HER2-downstream ERK at the transcriptional level. Another HER2-downstream kinase, Akt, was found to phosphorylate KRT19 on Ser35 and induce membrane translocation of KRT19 and remodeling of KRT19 from filamentous to granulous form. KRT19 phosphorylated by Akt could bind HER2 on the plasma membrane and stabilized HER2 via inhibition of proteasome-mediated degradation of HER2. Silencing of KRT19 by shRNA resulted in increased ubiquitination and destabilization of HER2. Moreover, treatment of KRT19 antibody resulted in downregulation of HER2 and reduced cell viability. These data provide a new rationale for targeting HER2-positive breast cancers.
Collapse
|
13
|
Fukuda M. Rab27 effectors, pleiotropic regulators in secretory pathways. Traffic 2013; 14:949-63. [PMID: 23678941 DOI: 10.1111/tra.12083] [Citation(s) in RCA: 167] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2013] [Revised: 05/13/2013] [Accepted: 05/16/2013] [Indexed: 12/18/2022]
Abstract
Rab27, a member of the small GTPase Rab family, is widely conserved in metazoan, and two Rab27 isoforms, Rab27A and Rab27B, are present in vertebrates. Rab27A was the first Rab protein whose dysfunction was found to cause a human hereditary disease, type 2 Griscelli syndrome, which is characterized by silvery hair and immunodeficiency. The discovery in the 21st century of three distinct types of mammalian Rab27A effectors [synaptotagmin-like protein (Slp), Slp homologue lacking C2 domains (Slac2), and Munc13-4] that specifically bind active Rab27A has greatly accelerated our understanding not only of the molecular mechanisms of Rab27A-mediated membrane traffic (e.g. melanosome transport and regulated secretion) but of the symptoms of Griscelli syndrome patients at the molecular level. Because Rab27B is widely expressed in various tissues together with Rab27A and has been found to have the ability to bind all of the Rab27A effectors that have been tested, Rab27A and Rab27B were initially thought to function redundantly by sharing common Rab27 effectors. However, recent evidence has indicated that by interacting with different Rab27 effectors Rab27A and Rab27B play different roles in special types of secretion (e.g. exosome secretion and mast cell secretion) even within the same cell type. In this review article, I describe the current state of our understanding of the functions of Rab27 effectors in secretory pathways.
Collapse
Affiliation(s)
- Mitsunori Fukuda
- Laboratory of Membrane Trafficking Mechanisms, Department of Developmental Biology and Neurosciences, Graduate School of Life Sciences, Tohoku University, Aobayama, Aoba-ku, Sendai, Miyagi 980-8578, Japan.
| |
Collapse
|
14
|
Catz SD. Regulation of vesicular trafficking and leukocyte function by Rab27 GTPases and their effectors. J Leukoc Biol 2013; 94:613-22. [PMID: 23378593 DOI: 10.1189/jlb.1112600] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The Rab27 family of GTPases regulates the efficiency and specificity of exocytosis in hematopoietic cells, including neutrophils, CTLs, NK cells, and mast cells. However, the mechanisms regulated by Rab27 GTPases are cell-specific, as they depend on the differential expression and function of particular effector molecules that are recruited by the GTPases. In addition, Rab27 GTPases participate in multiple steps of the regulation of the secretory process, including priming, tethering, docking, and fusion through sequential interaction with multiple effector molecules. Finally, recent reports suggest that Rab27 GTPases and their effectors regulate vesicular trafficking mechanisms other than exocytosis, including endocytosis and phagocytosis. This review focuses on the latest discoveries on the function of Rab27 GTPases and their effectors Munc13-4 and Slp1 in neutrophil function comparatively to their functions in other leukocytes.
Collapse
Affiliation(s)
- Sergio Daniel Catz
- 1.The Scripps Research Institute, 10550 North Torrey Pines Rd., La Jolla, CA 92037, USA. ; Twitter: http://www.scripps.edu/catz/
| |
Collapse
|
15
|
Li J, Song J, Cassidy MG, Rychahou P, Starr ME, Liu J, Li X, Epperly G, Weiss HL, Townsend CM, Gao T, Evers BM. PI3K p110α/Akt signaling negatively regulates secretion of the intestinal peptide neurotensin through interference of granule transport. Mol Endocrinol 2012; 26:1380-93. [PMID: 22700584 DOI: 10.1210/me.2012-1024] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Neurotensin (NT), an intestinal peptide secreted from N cells in the small bowel, regulates a variety of physiological functions of the gastrointestinal tract, including secretion, gut motility, and intestinal growth. The class IA phosphatidylinositol 3-kinase (PI3K) family, which comprised of p110 catalytic (α, β and δ) and p85 regulatory subunits, has been implicated in the regulation of hormone secretion from endocrine cells. However, the underlying mechanisms remain poorly understood. In particular, the role of PI3K in intestinal peptide secretion is not known. Here, we show that PI3K catalytic subunit, p110α, negatively regulates NT secretion in vitro and in vivo. We demonstrate that inhibition of p110α, but not p110β, induces NT release in BON, a human endocrine cell line, which expresses NT mRNA and produces NT peptide in a manner analogous to N cells, and QGP-1, a pancreatic endocrine cell line that produces NT peptide. In contrast, overexpression of p110α decreases NT secretion. Consistently, p110α-inhibition increases plasma NT levels in mice. To further delineate the mechanisms contributing to this effect, we demonstrate that inhibition of p110α increases NT granule trafficking by up-regulating α-tubulin acetylation; NT secretion is prevented by overexpression of HDAC6, an α-tubulin deacetylase. Moreover, ras-related protein Rab27A (a small G protein) and kinase D-interacting substrate of 220 kDa (Kidins220), which are associated with NT granules, play a negative and positive role, respectively, in p110α-inhibition-induced NT secretion. Our findings identify the critical role and novel mechanisms for the PI3K signaling pathway in the control of intestinal hormone granule transport and release.
Collapse
Affiliation(s)
- Jing Li
- Department of Surgery, University of Kentucky, Lexington, Kentucky, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Johnson JL, Monfregola J, Napolitano G, Kiosses WB, Catz SD. Vesicular trafficking through cortical actin during exocytosis is regulated by the Rab27a effector JFC1/Slp1 and the RhoA-GTPase-activating protein Gem-interacting protein. Mol Biol Cell 2012; 23:1902-16. [PMID: 22438581 PMCID: PMC3350554 DOI: 10.1091/mbc.e11-12-1001] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
The mechanism of cytoskeleton remodeling during exocytosis is not well defined. A combination of vesicular dynamics and functional studies shows that the Rab27a effector JFC1 and the RhoA-GTPase–activating protein Gem-interacting protein are necessary for RhoA regulation, actin depolymerization, and vesicular transport through the actin cortex during exocytosis. Cytoskeleton remodeling is important for the regulation of vesicular transport associated with exocytosis, but a direct association between granular secretory proteins and actin-remodeling molecules has not been shown, and this mechanism remains obscure. Using a proteomic approach, we identified the RhoA-GTPase–activating protein Gem-interacting protein (GMIP) as a factor that associates with the Rab27a effector JFC1 and modulates vesicular transport and exocytosis. GMIP down-regulation induced RhoA activation and actin polymerization. Importantly, GMIP-down-regulated cells showed impaired vesicular transport and exocytosis, while inhibition of the RhoA-signaling pathway induced actin depolymerization and facilitated exocytosis. We show that RhoA activity polarizes around JFC1-containing secretory granules, suggesting that it may control directionality of granule movement. Using quantitative live-cell microscopy, we show that JFC1-containing secretory organelles move in areas near the plasma membrane deprived of polymerized actin and that dynamic vesicles maintain an actin-free environment in their surroundings. Supporting a role for JFC1 in RhoA inactivation and actin remodeling during exocytosis, JFC1 knockout neutrophils showed increased RhoA activity, and azurophilic granules were unable to traverse cortical actin in cells lacking JFC1. We propose that during exocytosis, actin depolymerization commences near the secretory organelle, not the plasma membrane, and that secretory granules use a JFC1- and GMIP-dependent molecular mechanism to traverse cortical actin.
Collapse
Affiliation(s)
- Jennifer L Johnson
- Department of Molecular and Experimental Medicine, Scripps Research Institute, La Jolla, CA 92037, USA
| | | | | | | | | |
Collapse
|
17
|
Wu SR, Cheng TS, Chen WC, Shyu HY, Ko CJ, Huang HP, Teng CH, Lin CH, Johnson MD, Lin CY, Lee MS. Matriptase is involved in ErbB-2-induced prostate cancer cell invasion. THE AMERICAN JOURNAL OF PATHOLOGY 2010; 177:3145-58. [PMID: 20971737 DOI: 10.2353/ajpath.2010.100228] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Deregulation of both ErbB-2 signaling and matriptase activity has been associated with human prostate cancer (PCa) progression. In this communication, we investigated the roles of both ErbB-2 signaling in matriptase zymogen activation and matriptase in ErbB-2-induced PCa malignancy. In a human PCa cell progression model, we observed that advanced PCa C-81 LNCaP cells exhibited an aggressive phenotype with increased cell migration and invasion capacity; these cells concurrently showed both enhanced ErbB-2 phosphorylation and increased matriptase zymogen activation compared with parental C-33 LNCaP cells. Moreover, ErbB2 activation, both ligand-dependent (eg, epidermal growth factor treatment) and ligand-independent (eg, overexpression), was able to induce matriptase zymogen activation in this cell line. Inhibition of ErbB-2 activity by either the specific inhibitor, AG825, in epidermal growth factor-treated C-33 LNCaP cells or ErbB-2 knockdown in C-81 LNCaP cells, reduced matriptase activation. These observations were confirmed by similar studies using both DU145 and PC3 cells. Together, these data suggest that ErbB-2 signaling plays an important role in matriptase zymogen activation. ErbB-2-enhanced matriptase activation was suppressed by a phosphatidylinositol 3-kinase inhibitor (ie, LY294002) but not by a MEK inhibitor (ie, PD98059). Suppression of matriptase expression by small hairpin RNA knockdown in ErbB-2-overexpressing LNCaP cells dramatically suppressed cancer cell invasion. In summary, our data indicate that ErbB-2 signaling via the phosphatidylinositol 3-kinase pathway results in up-regulated matriptase zymogen activity, which contributes to PCa cell invasion.
Collapse
Affiliation(s)
- Shang-Ru Wu
- Department of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, R817, 8F, No. 1, Section 1, Jen-Ai Rd, Taipei, Taiwan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Catz SD. Characterization of Rab27a and JFC1 as constituents of the secretory machinery of prostate-specific antigen in prostate carcinoma cells. Methods Enzymol 2008; 438:25-40. [PMID: 18413239 PMCID: PMC11960417 DOI: 10.1016/s0076-6879(07)38003-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/18/2023]
Abstract
Prostate-specific antigen (PSA) and prostate-specific acid phosphatase (PSAP) are produced by prostate carcinoma cells. Their secretion has implications in both prostate cancer diagnosis and progression. The mechanisms involved in PSA and PSAP secretion in response to androgens have remained relatively unknown. The small GTPase Rab27a regulates exocytosis in several tissues. Here, we present methods for the characterization of Rab27a and its effector JFC1/Slp1 as key components of the secretory machinery that regulates exocytosis in prostate carcinoma cells.
Collapse
Affiliation(s)
- Sergio D Catz
- Division of Biochemistry, Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, California, USA
| |
Collapse
|
19
|
Fang D, Hawke D, Zheng Y, Xia Y, Meisenhelder J, Nika H, Mills GB, Kobayashi R, Hunter T, Lu Z. Phosphorylation of beta-catenin by AKT promotes beta-catenin transcriptional activity. J Biol Chem 2007; 282:11221-9. [PMID: 17287208 PMCID: PMC1850976 DOI: 10.1074/jbc.m611871200] [Citation(s) in RCA: 709] [Impact Index Per Article: 39.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Increased transcriptional activity of beta-catenin resulting from Wnt/Wingless-dependent or -independent signaling has been detected in many types of human cancer, but the underlying mechanism of Wnt-independent regulation is poorly understood. We have demonstrated that AKT, which is activated downstream from epidermal growth factor receptor signaling, phosphorylates beta-catenin at Ser552 in vitro and in vivo. AKT-mediated phosphorylation of beta-catenin causes its disassociation from cell-cell contacts and accumulation in both the cytosol and the nucleus and enhances its interaction with 14-3-3zeta via a binding motif containing Ser552. Phosphorylation of beta-catenin by AKT increases its transcriptional activity and promotes tumor cell invasion, indicating that AKT-dependent regulation of beta-catenin plays a critical role in tumor invasion and development.
Collapse
Affiliation(s)
- Dexing Fang
- Brain Tumor Center and Department of Neuro-Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030
| | - David Hawke
- Department of Molecular Pathology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030
| | - Yanhua Zheng
- Brain Tumor Center and Department of Neuro-Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030
| | - Yan Xia
- Brain Tumor Center and Department of Neuro-Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030
| | - Jill Meisenhelder
- Molecular and Cell Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, California 92037
| | - Heinz Nika
- Department of Molecular Pathology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030
| | - Gordon B. Mills
- Department of Systems Biology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030
| | - Ryuji Kobayashi
- Department of Molecular Pathology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030
| | - Tony Hunter
- Molecular and Cell Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, California 92037
| | - Zhimin Lu
- Brain Tumor Center and Department of Neuro-Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030
- Department of Molecular Genetics, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030
- The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, Texas 77030
| |
Collapse
|
20
|
Munafó D, Johnson J, Ellis B, Rutschmann S, Beutler B, Catz S. Rab27a is a key component of the secretory machinery of azurophilic granules in granulocytes. Biochem J 2007; 402:229-39. [PMID: 17090228 PMCID: PMC1798439 DOI: 10.1042/bj20060950] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Neutrophils kill micro-organisms using microbicidal products that they release into the phagosome or into the extracellular space. The secretory machinery utilized by neutrophils is poorly characterized. We show that the small GTPase Rab27a is an essential component of the secretory machinery of azurophilic granules in granulocytes. Rab27a-deficient mice have impaired secretion of MPO (myeloperoxidase) into the plasma in response to lipopolysaccharide. Cell fractionation analysis revealed that Rab27a and the Rab27a effector protein JFC1/Slp1 (synaptotagmin-like protein 1) are distributed principally in the low-density fraction containing a minor population of MPO-containing granules. By immunofluorescence microscopy, we detected Rab27a and JFC1/Slp1 in a minor subpopulation of MPO-containing granules. Interference with the JFC1/Slp1-Rab27a secretory machinery impaired secretion of MPO in permeabilized neutrophils. The expression of Rab27a was dramatically increased when promyelocytic HL-60 cells were differentiated into granulocytes but not when they were differentiated into monocytes. Down-regulation of Rab27a in HL-60 cells by RNA interference did not affect JFC1/Slp1 expression but significantly decreased the secretion of MPO. Neither Rab27a nor JFC1/Slp1 was integrated into the phagolysosome membrane during phagocytosis. Neutrophils from Rab27a-deficient mice efficiently phagocytose zymosan opsonized particles and deliver MPO to the phagosome. We conclude that Rab27a and JFC1/Slp1 permit MPO release into the surrounding milieu and constitute key components of the secretory machinery of azurophilic granules in granulocytes. Our results suggest that the granules implicated in cargo release towards the surrounding milieu are molecularly and mechanistically different from those involved in their release towards the phagolysosome.
Collapse
Affiliation(s)
- Daniela B. Munafó
- *Department of Molecular and Experimental Medicine, Division of Biochemistry, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla CA, 92037, U.S.A
| | - Jennifer L. Johnson
- *Department of Molecular and Experimental Medicine, Division of Biochemistry, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla CA, 92037, U.S.A
| | - Beverly A. Ellis
- *Department of Molecular and Experimental Medicine, Division of Biochemistry, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla CA, 92037, U.S.A
| | - Sophie Rutschmann
- †Department of Immunology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, U.S.A
| | - Bruce Beutler
- †Department of Immunology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, U.S.A
| | - Sergio D. Catz
- *Department of Molecular and Experimental Medicine, Division of Biochemistry, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla CA, 92037, U.S.A
- To whom correspondence should be addressed (email )
| |
Collapse
|
21
|
Hattula K, Furuhjelm J, Tikkanen J, Tanhuanpää K, Laakkonen P, Peränen J. Characterization of the Rab8-specific membrane traffic route linked to protrusion formation. J Cell Sci 2006; 119:4866-77. [PMID: 17105768 DOI: 10.1242/jcs.03275] [Citation(s) in RCA: 177] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Rab8 has a drastic effect on cell shape, but the membrane trafficking route it regulates is poorly defined. Here, we show that endogenous and ectopically expressed Rab8 is associated with macropinosomes generated at ruffling membrane domains. These macropinosomes fuse or transform into tubules that move toward the cell center, from where they are recycled back to the leading edge. The biogenesis of these tubules is dependent on actin and microtubular dynamics. Expression of dominant-negative Rab8 mutants or depletion of Rab8 by RNA interference inhibit protrusion formation, but promote cell-cell adhesion and actin stress fiber formation, whereas expression of the constitutively active Rab8-Q67L has the opposite effect. Rab8 localization overlaps with both Rab11 and Arf6, and is functionally linked to Arf6. We also demonstrate that Rab8 activity is needed for the transport of transferrin and the transferrin receptor to the pericentriolar region and to cell protrusions, and that Rab8 controls the traffic of cholera toxin B to the Golgi compartment. Finally, Rab8 colocalizes and binds specifically to a synaptotagmin-like protein (Slp1/JFC1), which is involved in controlling Rab8 membrane dynamics. We propose that Rab8 regulates a membrane-recycling pathway that mediates protrusion formation.
Collapse
Affiliation(s)
- Katarina Hattula
- Institute of Biotechnology, PO Box 56 (Viikinkaari 9), FIN-00014 University of Helsinki, Finland
| | | | | | | | | | | |
Collapse
|
22
|
Johnson J, Ellis B, Noack D, Seabra M, Catz S. The Rab27a-binding protein, JFC1, regulates androgen-dependent secretion of prostate-specific antigen and prostatic-specific acid phosphatase. Biochem J 2005; 391:699-710. [PMID: 16004602 PMCID: PMC1276972 DOI: 10.1042/bj20050380] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2005] [Revised: 06/29/2005] [Accepted: 07/08/2005] [Indexed: 11/17/2022]
Abstract
Two of the major proteins secreted by the prostate epithelium secretory cells are PSA (prostate-specific antigen) and PSAP (prostatic-specific acid phosphatase). The molecules involved in the secretory machinery of PSA and PSAP, and the regulation of this machinery, remain unknown. In the present paper, we provide evidence that JFC1 [synaptotagmin-like protein (slp1)], a Rab27a- and PtdIns(3,4,5)P3-binding protein, regulates the androgen-dependent secretion of PSAP and PSA in human LNCaP prostate carcinoma cells. Androgen-dependent PSAP secretion was significantly inhibited in cells that expressed the C2A domain of JFC1 [PtdIns(3,4,5)P3-binding-domain], but was unaffected by JFC1 overexpression. Conversely, PSA secretion was not inhibited by the C2A domain of JFC1. We show, using immunofluorescence analysis, that JFC1 co-localizes with PSAP, but rarely with PSA, in prostate granules, suggesting that JFC1 is part of the PSAP secretory machinery. However, PSA secretion was significantly increased in LNCaP cells that overexpressed JFC1, indicating that the secretion of PSA is susceptible to variations in the intracellular concentration of JFC1. Both PSAP and PSA secretion was increased by overexpression of wild-type Rab27a or the constitutively active Rab27aQ78L. The secretion of PSA was partially inhibited in the presence of LY294002, while the secretion of PSAP was completely abolished by the PI3K (phosphoinositide 3-kinase) inhibitor. This supports the view that PI3K plays a differential role in the secretion of prostate secretory markers. In conclusion, we present evidence that JFC1 differentially regulates the secretion of PSAP and PSA, and that Rab27a and PI3K play a central role in the exocytosis of prostate-specific markers.
Collapse
Key Words
- exocytosis
- phosphoinositide 3-kinase (pi3k)
- prostate-specific antigen (psa)
- prostatic-specific acid phosphatase (psap)
- synaptotagmin-like protein
- vesicular trafficking
- dsred, red fluorescent protein from discosoma sp.
- eea1, early endosome antigen 1
- egfp, enhanced green fluorescent protein
- nf-κb, nuclear factor κb
- pi3k, phosphoinositide 3-kinase
- pip3, ptdins(3,4,5)p3
- psa, prostate-specific antigen
- psap, prostate-specific acid phosphatase
- slp, synaptotagmin-like protein
- syt, synaptotagmin
- t-snare, target-associated soluble n-ethylmaleimide-sensitive fusion protein attachment protein receptor
- vamp-2, vesicle-associated membrane protein-2
Collapse
Affiliation(s)
- Jennifer L. Johnson
- *Division of Biochemistry, Department of Molecular and Experimental Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, U.S.A
| | - Beverly A. Ellis
- *Division of Biochemistry, Department of Molecular and Experimental Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, U.S.A
| | - Deborah Noack
- *Division of Biochemistry, Department of Molecular and Experimental Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, U.S.A
| | - Miguel C. Seabra
- †Cell and Molecular Biology Section, Division of Biomedical Sciences, Faculty of Medicine, Imperial College London, London SW7 2AZ, U.K
| | - Sergio D. Catz
- *Division of Biochemistry, Department of Molecular and Experimental Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, U.S.A
| |
Collapse
|